6,174
Views
52
CrossRef citations to date
0
Altmetric
Review Article

Optimizing antifungal choice and administration

Pages 13-18 | Accepted 11 Dec 2012, Published online: 26 Apr 2013

Abstract

Background:

The antifungal armamentarium includes a number of drug classes and agents within each class. Successful IFI management depends on optimal matching of drug choice with the individual patient and causative pathogen, and maximizing effectiveness of the selected drug through appropriate dosing and toxicity management.

Objective:

This review is intended to provide a brief overview of key factors involved in optimizing antifungal choice and administration for patients with invasive fungal infections (IFIs).

Findings:

Antifungals differ in spectrum of activity, and these differences are critical when selecting the antifungal most likely to provide success for a patient with an IFI. When the species has not yet been identified, an analysis of regional epidemiology and risk factors can provide clues as to the most likely pathogen. For severely immunocompromised patients, a fungicidal agent may be preferred over a fungistatic agent, although more research is needed in this area. Triazoles, particularly itraconazole and posaconazole, exhibit great interpatient pharmacokinetic variability related to absorption. Steps can be taken to maximize absorption when using these agents. Voriconazole concentration is affected by polymorphisms in the major metabolic enzyme, cytochrome P450 2C19. Triazoles, and to a lesser extent other antifungals, are also subject to drug–drug interactions, which needs to be considered when selecting a particular antifungal agent for use in a severely ill patient on polypharmacy. Therapeutic drug monitoring may be a useful adjunct for patients receiving itraconazole, voriconazole, or posaconazole. When the IFI involves a pharmacologically protected site, such as the central nervous system (CNS) or eye, 5-fluorocytosine, fluconazole, or voriconazole are generally preferred. Echinocandin penetration is typically inadequate for IFIs of the CNS or eye. Antifungal agents also differ in their toxicity profiles, and these issues also need to be considered and managed when making an antifungal choice.

Conclusion:

Successful management of IFIs relies in part on the accurate selection of an antifungal agent for the infection. Drug characteristics can help in the selection of drug therapy. These characteristics include the drug’s spectrum of activity, pharmacokinetics, pharmacodynamics, toxicity profile, and distribution to the infection site. Matching the drug profile to the patient and fungal species contribute to optimal management of infection.

Introduction

Clinicians now have multiple drugs from several antifungal drug classes for the management of invasive fungal infections (IFIs). These include four drugs of the polyene class (amphotericin B [AmB] deoxycholate and three lipid-based amphotericin B formulations: amphotericin B colloidal dispersion [ABCD], amphotericin B lipid complex [ABLC], and liposomal AmB [L-AMB]); four triazole drugs (itraconazole, fluconazole, voriconazole, and posaconazole); three echinocandins (caspofungin, micafungin, and anidulafungin), and 5-fluorocytosine (5FC, also known as flucytosine), a pyrimidine analogCitation1,Citation2. Additional agents are in developmentCitation3,Citation4.

The azoles are available as either intravenous (IV) or oral formulations (with the exception of posaconazole, which does not currently have an IV formulation, and itraconazole, which has an IV formulation in Europe but not the United States); the polyenes and echinocandins can only be administered parenterally; and 5FC is only available in an oral preparationCitation5. Besides route of administration, other variables differentiate antifungal agents, and can be important when selecting the most suitable agent.

This review examines how to utilize these differentiating characteristics to optimize antifungal choice and administration for patients. Ideally, data from randomized controlled, head-to-head clinical trials would be available when selecting an agent (e.g., Herbrecht et al., comparing voriconazole and AmB for invasive aspergillosis [IA]Citation6), but often these data are not available. Hence, an understanding of drug characteristics assumes greater importance for drug selection.

Spectrum of activity

A critically important consideration is the agent’s spectrum of activity. Antifungal classes, and even agents within the same class, often exhibit differential activity against particular fungal pathogens (Citation1,Citation5,Citation7,Citation8). For example, different antifungals show variable activity against different Candida spp. C. albicans and C. tropicalis are susceptible to all agents, but triazoles show minimal or variable activity against C. glabrata, and echinocandins exhibit the greatest activityCitation1,Citation5,Citation7,Citation8. Both polyenes and (particularly) echinocandins are active against C. krusei, while only voriconazole and to a lesser extent posaconazole exhibit reasonable activityCitation1,Citation5,Citation7–9. Polyenes and triazoles demonstrate good activity for C. parapsilosis, while echinocandins are less active against this pathogen. Polyenes are not effective for C. lusitaniae. The echinocandins exhibit reduced activity against C. parapsilosis and C. guilliermondii.

Table 1.  Antifungal spectrum of activity against more common invasive fungal pathogens (data from Lewis, 2011Citation1; Denning and Hope, 2010Citation5; Dodds Ashley et al., 2006Citation7; and Pfaller and Diekema, 2010Citation8).

Although important, identification at the species level is often not available in most microbiology laboratories until several days into the course of treatment; delayed or inadequate initial antifungal therapy has been linked with poorer outcomesCitation10–14. Hence, ‘hitting the target right the first time’ requires an understanding of local epidemiology patterns, and using this in conjunction with antifungal spectrum of activity to guide initial drug selection. Candida spp. distribution and patterns of resistance can vary not only by clinical serviceCitation8 but also globally and by geographic regionsCitation15–20. In the United States, the most recent survey has demonstrated the continuing emergence of C. glabrata and corresponding decline in C. albicans in invasive candidiasis (IC)Citation21. Therefore, at my institution, the University of Wisconsin Hospital in Madison, echinocandins are first-line therapy for IC while waiting for species identification.

Species identification has also become important for Aspergillus and other pathogenic molds. A. fumigatus – the most common cause of invasive aspergillosis (IA) – is susceptible to polyenes, echinocandins, and all triazoles except fluconazole, while A. terreus and A. nidulans are susceptible to triazoles (except fluconazole) and echinocandins but not polyenesCitation5,Citation7,Citation8,Citation22–24. Most Mucor spp. or Zygomycetes are susceptible to posaconazole, but not to echinocandins, voriconazole, or other azoles; polyenes exhibit variable activity against these pathogensCitation1,Citation5,Citation7,Citation8,Citation25. For Fusarium spp. and Scedosporium spp., only voriconazole and posaconazole exhibit relatively consistent activity against these pathogens, with polyenes exhibiting more variable activity for Fusarium but limited activity for Scedosporium spp. Neither echinocandins nor other triazoles are active against any of these speciesCitation5,Citation7,Citation8.

Cryptococcus neoformans is susceptible to polyenes, 5FC, all the triazoles, but not echinocandinsCitation1,Citation5,Citation7,Citation8,Citation24,Citation26. The fungi that cause histoplasmosis, blastomycosis, and coccidioidomycosis exhibit in vitro susceptibility to polyenes and all triazolesCitation1,Citation7,Citation8.

Pharmacokinetics (PK) and therapeutic drug monitoring (TDM)

PK issues are also important considerations for drug selection and when trying to optimize drug levels for maximal benefit and minimal risk. PK variability is a particular concern with triazole therapy, where various factors affect absorption and bioavailability and where genetic polymorphisms of cytochrome P450 (CYP) 2C19 can alter voriconazole plasma levels. Drug–drug interactions and comorbidities (renal or hepatic insufficiency) can also have important PK effects.

Oral itraconazole and posaconazole show greater variability in absorption than oral fluconazole or voriconazole. The capsular form of itraconazole is associated with wide variabilityCitation27–29, with markedly lower absorption and plasma concentrations when administered under fasting versus fed conditionsCitation30,Citation31. Itraconazole solution demonstrates more predictable absorption and higher plasma concentrations compared to the itraconazole capsule formulation, particularly under fasting conditionsCitation32,Citation33. Administration of itraconazole capsules with an acidic beverage has been shown to enhance absorption. Food also increases the bioavailability of the capsule formCitation34. Wide interpatient pharmacokinetic variability has also been observed with posaconazoleCitation35, and similar to itraconazole, absorption and bioavailability of posaconazole is markedly increased when administered with food or a nutritional supplement versus when fastingCitation36–39. Gastric acidity is also important for posaconazole absorptionCitation40. Finally, posaconazole absorption is saturable, thus despite a long elimination half life, exposure is increased when administered as divided doses of 200 mg four times daily versus as a single 800 mg daily doseCitation41,Citation42. Interpatient variability with voriconazole is primarily due to differences in metabolism related to CYP2C19 polymorphisms, rather than absorption issues, although voriconazole absorption is modestly reduced by administration with foodCitation43. CYP2C19 is the major determinant of voriconazole metabolism and clearance, and patients who are homozygous extensive CYP2C19 metabolizers have markedly lower plasma concentrations than homozygous poor metabolizers or heterozygous extensive metabolizersCitation44,Citation45. Fluconazole demonstrates predictable, linear PK that is unaffected by food or gastric acidityCitation46–48.

TDM has been proposed as a potential tool for optimizing therapy with itraconazole, voriconazole, and posaconazoleCitation35,Citation49–52. Each of these drugs meet widely accepted criteria for TDM: unpredictable population PK, a relatively narrow therapeutic window, and a fairly well defined clinical therapeutic rangeCitation49,Citation51. TDM recommended trough concentrations for voriconazole and posaconazole efficacy are each approximately >1 μg/mLCitation50,Citation51,Citation53–57. A recommended trough <5 μg/mL has been recommended to limit the likelihood of voriconazole toxicity; a toxicity trough associated with posaconazole has not been definedCitation51.

Comorbid organ dysfunction, specifically renal or hepatic insufficiency, alters the PK of certain antifungals and impacts either drug selection or the dosing regimen design. Dose reductions of fluconazole or 5FC are recommended for patients with renal sufficiency. No dosing adjustments for any of the polyene formulations, oral voriconazole, posaconazole, or any of the echinocandins are required in patients with renal dysfunctionCitation7. However, caution should be exercised when IV voriconazole is used in patients with a creatinine clearance (CrCl) <50 mL/min or with IV itraconazole in patients with CrCl <30 mL/minCitation7. This is because the cyclodextrin component of these parenteral formulations can accumulate in patients with renal disease, and some animal studies suggest very high cyclodextrin may be associated with renal toxicityCitation58. Voriconazole dose reduction is recommendations for patients with mild-to-moderate hepatic dysfunctionCitation59. Similarly caspofungin dose should be halved in patients with moderate hepatic insufficiencyCitation60. No dosing adjustments are needed based on hepatic insufficiency for any other antifungalCitation7.

Drug interactions and toxicity

The likelihood for drug–drug interactions is greater with triazoles than other major antifungal classes. All the triazoles are substrates for one or more CYP450 enzymes, which creates the potential for bidirectional drug–drug interactions when these drugs are used concomitantly with other drugs that are also substrates for these enzymes. All triazoles are either moderate or strong inhibitors of CYP3A4, while fluconazole, and voriconazole also inhibit CYP2C9Citation1. Voriconazole is also a strong inhibitor of CYP2C19Citation1. Because of this, clinically relevant potential drug–drug interactions can occur when particular triazoles are coadministered with drugs that either induce or inhibit CYP450 isoenzymes or that are substrates for them. A large list of significant drug–drug interactions have been noted with triazoles (although less with posaconazole)Citation61,Citation62.

Drug–drug interactions need to be considered not only when initiating antifungal therapy, but also when discontinuing it. One will often need to adjust the dose of the remaining non-antifungal drug. Interestingly, at our institution, we sometimes use the voriconazole-elevating effects of coadministered omeprazoleCitation63 to increase voriconazole exposure in situations where simple dosage increases have minimal impact.

Drug–drug interactions involving other (non-triazole) antifungals have been reviewed elsewhereCitation7,Citation64–66. Echinocandins are not appreciable substrates, inducers, or inhibitors of CYP450 isoenzymes, and exhibit few clinically relevant drug–drug interactions. Clinically relevant polyene drug–drug interactions are not associated with drug exposure per se, but rather with their polyene nephrotoxicity, additive nephrotoxic effects when coadministered with other nephrotoxic agents, and altered renal elimination or electrolyte imbalances due to these renal effects.

Toxicities associated with antifungal agents can also impact drug choice and monitoringCitation1,Citation2,Citation5,Citation7,Citation64,Citation67. Toxicity associated with 5FC includes hepatitis and myelosuppression, especially at serum concentrations >100 μg/mL. Polyene antifungals exhibit dose-related nephrotoxicity which is reduced with newer lipid formulations of AmB compared to conventional AmB deoxycholateCitation2,Citation68. Polyenes have also been linked with common infusion-related reactions and less frequently anemia. Furthermore, polyene-related nephrotoxicity increases the risk of serious 5FC-related adverse events such as bone marrow suppression when polyenes are coadministered with this 5FCCitation69,Citation70. Triazoles are generally safe and well toleratedCitation2,Citation68. The most common side effects are gastrointestinal. One important consideration with the triazole class is modest QTc prolongation. This is not typically important unless they are combined with other pharmacologic agents with similar conduction properties. Among the class, voriconazole is associated with greater but still low incidence of hepatotoxicity and also exhibits several unique side effects including visual disturbances and photosensitivityCitation71. Very high concentrations of voriconazole have also been associated with reversible neurologic toxicity, with 31% (5/16) of patients with trough levels >5.5 mg/L experiencing encephalopathy as a serious adverse event in one study, and none of 30 patients with trough levels ≤5.5 mg/LCitation53. Onset of encephalopathy ranged from 5 to 30 days following start of voriconazole therapy. Fluconazole is a well tolerated antifungal without significant toxicity. All triazoles are also teratogenic. Echinocandins are generally well tolerated and exhibit few toxicities, the most common being occasional mild infusion-related reactions.

Distribution to infection sites

Distribution to the site of infection is another PK variable of potential importance when individualizing antifungal therapy. A number of pathogenic fungi have a propensity to disseminate to pharmacologically protected body sites – particularly the central nervous system (CNS) or eye, or urine with Candida spp. – and antifungals differ in their ability to penetrate these tissuesCitation72. Of the triazoles, only fluconazole and voriconazole penetrate to any measurable degree into the CSF or intra-ocular compartments, while the echinocandins typically lack penetration or achieve very low levels in these tissuesCitation73,Citation74. 5FC exhibits excellent penetration to all common infection sites, while polyenes show only modest penetration into the CSF. Fluconazole and 5FC are the only antifungals that demonstrate any appreciable excretion into the urine for patients with asymptomatic Candida urinary tract infectionsCitation75.

Conclusions

Optimal IFI management depends on multiple factors, including rapid and accurate diagnosis and early treatment with an antifungal agent or antifungal combination best suited for the individual patient and causative pathogen. Once a suitable antifungal is selected, it is important to maximize effectiveness by managing its administration and taking the appropriate steps to maximize efficacy and reduce toxicity. Under certain circumstances, TDM may be useful.

Knowledge of the agent’s spectrum of activity is essential, particularly when the species has been identified, but epidemiology patterns and risk factor evaluation are important guides when species identification is not available. Certain antifungals and dosages are better than others when the IFI involves a pharmacologically protected site. When selecting certain triazoles, an understanding of factors affecting absorption and ways to optimize exposure is essential, including an understanding of drug–drug interactions. Toxicity monitoring is important when using polyenes and 5FC, and to a lesser extent with certain triazoles. Risk of nephrotoxicity is a concern when considering polyene selection. Knowledge of all these factors contributes to an optimal therapy regimen for patients with an IFI.

Transparency

Declaration of funding

This review was funded by an educational grant from Merck & Co. Inc.

Declaration of financial/other relationships

D.A. has disclosed that he has been a consultant to Astellas and Merck.

CMRO peer reviewers on this manuscript have received honoraria for their review work, but have no other relevant financial relationships to disclose.

Supplemental material

Supplementary Material

Download PDF (76.3 KB)

Acknowledgment

The authors of this supplement thank Global Education Exchange Inc. for editorial support.

References

  • Lewis RE. Current concepts in antifungal pharmacology. Mayo Clin Proc 2011;86:805-17
  • Pound MW, Townsend ML, Dimondi V, et al. Overview of treatment options for invasive fungal infections. Med Mycol 2011;49:561-80
  • Ostrosky-Zeichner L, Casadevall A, Galgiani JN, et al. An insight into the antifungal pipeline: selected new molecules and beyond. Nat Rev Drug Discov 2010;9:719-27
  • Pitman SK, Drew RH, Perfect JR. Addressing current medical needs in invasive fungal infection prevention and treatment with new antifungal agents, strategies and formulations. Expert Opin Emerging Drugs 2011;16:559-86
  • Denning DW, Hope WW. Therapy for fungal diseases: opportunities and priorities. Trends Microbiol 2010;18:195-204
  • Herbrecht R, Denning DW, Patterson TF, et al. Voriconazole versus amphotericin B for primary therapy of invasive aspergillosis. N Engl J Med 2002;347:408-15
  • Dodds Ashley ES, Lewis R, Lewis JS, et al. Pharmacology of systemic antifungal agents. Clin Infect Dis 2006;43:S28-39
  • Pfaller MA, Diekema DJ. Epidemiology of invasive mycoses in North America. Crit Rev Microbiol 2010;36:1-53
  • Pfaller MA, Messer SA, Moet GJ, et al. Candida bloodstream infections: comparison of species distribution and resistance to echinocandin and azole antifungal agents in Intensive Care Unit (ICU) and non-ICU settings in the SENTRY Antimicrobial Surveillance Program (2008–2009). Int J Antimicrob Agents 2011;38:65-9
  • Garey KW, Pai MP, Suda KJ, et al. Inadequacy of fluconazole dosing in patients with candidemia based on Infectious Diseases Society of America (IDSA) guidelines. Pharmacoepidemiol Drug Saf 2007;16:919-27
  • Garey KW, Rege M, Pai MP, et al. Time to initiation of fluconazole therapy impacts mortality in patients with candidemia: a multi-institutional study. Clin Infect Dis 2006;43:25-31
  • Garey KW, Turpin RS, Bearden DT, et al. Economic analysis of inadequate fluconazole therapy in non-neutropenic patients with candidaemia: a multi-institutional study. Int J Antimicrob Agents 2007;29:557-62
  • Nolla-Salas J, Sitges-Serra A, Leon-Gil C, et al. Candidemia in non-neutropenic critically ill patients: analysis of prognostic factors and assessment of systemic antifungal therapy. Study group of fungal infection in the ICU. Intensive Care Med 1997;23:23-30
  • Parkins MD, Sabuda DM, Elsayed S, et al. Adequacy of empirical antifungal therapy and effect on outcome among patients with invasive Candida species infections. J Antimicrob Chemother 2007;60:613-18
  • Jung SI, Shin JH, Song JH, et al. Multicenter surveillance of species distribution and antifungal susceptibilities of Candida bloodstream isolates in South Korea. Med Mycol 2010;48:669-74
  • Lortholary O, Desnos-Ollivier M, Sitbon K, et al. Recent exposure to caspofungin or fluconazole influences the epidemiology of candidemia: a prospective multicenter study involving 2441 patients. Antimicrob Agents Chemother 2011;55:532-8
  • Lyon GM, Karatela S, Sunay S, et al. Antifungal susceptibility testing of Candida isolates from the Candida surveillance study. J Clin Microbiol 2010;48:1270-5
  • Pfaller MA, Diekema DJ, Gibbs DL, et al. Geographic variation in the frequency of isolation and fluconazole and voriconazole susceptibilities of Candida glabrata: an assessment from the ARTEMIS DISK Global Antifungal Surveillance Program. Diagn Microbiol Infect Dis 2010;67:162-71
  • Pfaller MA, Moet GJ, Messer SA, et al. Geographic variations in species distribution and echinocandin and azole antifungal resistance rates among Candida bloodstream infection isolates: report from the SENTRY Antimicrobial Surveillance Program (2008 to 2009). J Clin Microbiol 2011;49:396-9
  • Tan TY, Tan AL, Tee NW, et al. A retrospective analysis of antifungal susceptibilities of Candida bloodstream isolates from Singapore hospitals. Ann Acad Med Singapore 2008;37:835-40
  • Lockhart SR, Iqbal N, Cleveland AA, et al. Species identification and antifungal susceptibility testing of Candida bloodstream isolates from population-based surveillance studies in two U.S. cities from 2008 to 2011. J Clin Microbiol. 2012;50:3435-42
  • Pfaller MA, Boyken L, Hollis RJ, et al. In vitro susceptibility of clinical isolates of Aspergillus spp. to anidulafungin, caspofungin, and micafungin: a head-to-head comparison using the CLSI M38-A2 broth microdilution method. J Clin Microbiol 2009;47:3323-5
  • Pfaller MA, Diekema DJ, Ghannoum MA, et al. Wild-type MIC distribution and epidemiological cutoff values for Aspergillus fumigatus and three triazoles as determined by the Clinical and Laboratory Standards Institute broth microdilution methods. J Clin Microbiol 2009;47:3142-6
  • Pfaller MA, Castanheira M, Messer SA, et al. Echinocandin and triazole antifungal susceptibility profiles for Candida spp., Cryptococcus neoformans, and Aspergillus fumigatus: application of new CLSI clinical breakpoints and epidemiologic cutoff values to characterize resistance in the SENTRY Antimicrobial Surveillance Program (2009). Diagn Microbiol Infect Dis 2011;69:45-50
  • Sun QN, Fothergill AW, McCarthy DI, et al. In vitro activities of posaconazole, itraconazole, voriconazole, amphotericin B, and fluconazole against 37 clinical isolates of zygomycetes. Antimicrob Agents Chemother 2002;46:1581-2
  • Pfaller MA, Castanheira M, Diekema DJ, et al. Wild-type MIC distributions and epidemiologic cutoff values for fluconazole, posaconazole, and voriconazole when testing Cryptococcus neoformans as determined by the CLSI broth microdilution method. Diagn Microbiol Infect Dis 2011;71:252-9
  • Hardin TC, Graybill JR, Fetchick R, et al. Pharmacokinetics of itraconazole following oral administration to normal volunteers. Antimicrob Agents Chemother 1988;32:1310-13
  • Lazo de la Vega S, Volkow P, Yeates RA, et al. Administration of the antimycotic agents fluconazole and itraconazole to leukaemia patients: a comparative pharmacokinetic study. Drugs Exp Clin Res 1994;20:69-75
  • Poirier JM, Berlioz F, Isnard F, et al. Marked intra- and inter-patient variability of itraconazole steady state plasma concentrations. Therapie 1996;51:163-7
  • Barone JA, Koh JG, Bierman RH, et al. Food interaction and steady-state pharmacokinetics of itraconazole capsules in healthy male volunteers. Antimicrob Agents Chemother 1993;37:778-84
  • Van Peer A, Woestenborghs R, Heykants J, et al. The effects of food and dose on the oral systemic availability of itraconazole in healthy subjects. Eur J Clin Pharmacol 1989;36:423-6
  • Cartledge JD, Midgely J, Gazzard BG. Itraconazole solution: higher serum drug concentrations and better clinical response rates than the capsule formulation in acquired immunodeficiency syndrome patients with candidosis. J Clin Pathol 1997;50:477-80
  • Van de Velde VJ, Van Peer AP, Heykants JJ, et al. Effect of food on the pharmacokinetics of a new hydroxypropyl-beta-cyclodextrin formulation of itraconazole. Pharmacotherapy 1996;16:424-8
  • Jaruratanasirikul S, Kleepkaew A. Influence of an acidic beverage (Coca-Cola) on the absorption of itraconazole. Eur J Clin Pharmacol 1997;52:235-7
  • Thompson GR 3rd, Rinaldi MG, Pennick G, et al. Posaconazole therapeutic drug monitoring: a reference laboratory experience. Antimicrob Agents Chemother 2009;53:2223-4
  • Courtney R, Radwanski E, Lim J, et al. Pharmacokinetics of posaconazole coadministered with antacid in fasting or nonfasting healthy men. Antimicrob Agents Chemother 2004;48:804-8
  • Krishna G, Ma L, Vickery D, et al. Effect of varying amounts of a liquid nutritional supplement on the pharmacokinetics of posaconazole in healthy volunteers. Antimicrob Agents Chemother 2009;53:4749-52
  • Krishna G, Moton A, Ma L, et al. Pharmacokinetics and absorption of posaconazole oral suspension under various gastric conditions in healthy volunteers. Antimicrob Agents Chemother 2009;53:958-66
  • Sansone-Parsons A, Krishna G, Calzetta A, et al. Effect of a nutritional supplement on posaconazole pharmacokinetics following oral administration to healthy volunteers. Antimicrob Agents Chemother 2006;50:1881-3
  • Jain R, Pottinger P. The effect of gastric acid on the absorption of posacaonzole [letter]. Clin Infect Dis 2008;46:1627
  • Ezzet F, Wexler D, Courtney R, et al. Oral bioavailability of posaconazole in fasted healthy subjects: comparison between three regimens and basis for clinical dosage recommendations. Clin Pharmacokinet 2005;44:211-20
  • Ullmann AJ, Cornely OA, Burchardt A, et al. Pharmacokinetics, safety, and efficacy of posaconazole in patients with persistent febrile neutropenia or refractory invasive fungal infection. Antimicrob Agents Chemother 2006;50:658-66
  • Purkins L, Wood N, Kleinermans D, et al. Effect of food on the pharmacokinetics of multiple-dose oral voriconazole. Br J Clin Pharmacol 2003;56(Suppl 1):17-23
  • Hyland R, Jones BC, Smith DA. Identification of the cytochrome P450 enzymes involved in the N-oxidation of voriconazole. Drug Metab Dispos 2003;31:540-7
  • Ikeda Y, Umemura K, Kondo K, et al. Pharmacokinetics of voriconazole and cytochrome P450 2C19 genetic status. Clin Pharmacol Ther 2004;75:587-8
  • Dodds-Ashley E. Management of drug and food interactions with azole antifungal agents in transplant recipients. Pharmacotherapy 2010;30:842-54
  • Zimmermann T, Yeates RA, Laufen H, et al. Influence of concomitant food intake on the oral absorption of two triazole antifungal agents, itraconazole and fluconazole. Eur J Clin Pharmacol 1994;46:147-50
  • Zimmermann T, Yeates RA, Riedel KD, et al. The influence of gastric pH on the pharmacokinetics of fluconazole: the effect of omeprazole. Int J Clin Pharmacol Ther 1994;32:491-6
  • Andes D, Pascual A, Marchetti O. Antifungal therapeutic drug monitoring: established and emerging indications. Antimicrob Agents Chemother 2009;53:24-34
  • Dolton MJ, Ray JE, Marriott D, et al. Posaconazole exposure–response relationship: evaluating the utility of therapeutic drug monitoring. Antimicrob Agents Chemother 2012;56:2806-13
  • Lewis RE. Antifungal drug monitoring. Curr Fungal Infect Rep 2010;4:158-67
  • Trifilio S, Pennick G, Pi J, et al. Monitoring plasma voriconazole levels may be necessary to avoid subtherapeutic levels in hematopoietic stem cell transplant recipients. Cancer 2007;109:1532-5
  • Pascual A, Calandra T, Bolay S, et al. Voriconazole therapeutic drug monitoring in patients with invasive mycoses improves efficacy and safety outcomes. Clin Infect Dis 2008;46:201-11
  • Smith J, Safdar N, Knasinski V, et al. Voriconazole therapeutic drug monitoring. Antimicrob Agents Chemother 2006;50:1570-2
  • Troke PF, Hockey HP, Hope WW. Observational study of the clinical efficacy of voriconazole and its relationship to plasma concentrations in patients. Antimicrob Agents Chemother 2011;55:4782-8
  • Jang SH, Colangelo PM, Gobburu JV. Exposure–response of posaconazole used for prophylaxis against invasive fungal infections: evaluating the need to adjust doses based on drug concentrations in plasma. Clin Pharmacol Ther 2010;88:115-19
  • Walsh TJ, Raad I, Patterson TF, et al. Treatment of invasive aspergillosis with posaconazole in patients who are refractory to or intolerant of conventional therapy: an externally controlled trial. Clin Infect Dis 2007;44:2-12
  • von Mach MA, Burhenne J, Weilemann LS. Accumulation of the solvent vehicle sulphobutylether beta cyclodextrin sodium in critically ill patients treated with intravenous voriconazole under renal replacement therapy. BMC Clin Pharmacol 2006;6:6
  • Weiler S, Zoller H, Graziadei I, et al. Altered pharmacokinetics of voriconazole in a patient with liver cirrhosis. Antimicrob Agents Chemother 2007;51:3459-60
  • Mistry GC, Migoya E, Deutsch PJ, et al. Single- and multiple-dose administration of caspofungin in patients with hepatic insufficiency: implications for safety and dosing recommendations. J Clin Pharmacol 2007;47:951-61
  • Bruggemann RJ, Alffenaar JW, Blijlevens NM, et al. Clinical relevance of the pharmacokinetic interactions of azole antifungal drugs with other coadministered agents. Clin Infect Dis 2009;48:1441-58
  • Gubbins PO. Triazole antifungal agents drug-drug interactions involving hepatic cytochrome P450. Expert Opin Drug Metab Toxicol 2011;7:1411-29
  • Wood N, Tan K, Purkins L, et al. Effect of omeprazole on the steady-state pharmacokinetics of voriconazole. Br J Clin Pharmacol 2003;56(Suppl 1):56-61
  • Chen SC, Slavin MA, Sorrell TC. Echinocandin antifungal drugs in fungal infections: a comparison. Drugs 2011;71:11-41
  • Gubbins PO, Heldenbrand S. Clinically relevant drug interactions of current antifungal agents. Mycoses 2010;53:95-113
  • Kontoyiannis DP. Invasive mycoses: strategies for effective management. Am J Med 2012;125:S25-38
  • Laniado-Laborin R, Cabrales-Vargas MN. Amphotericin B: side effects and toxicity. Rev Iberoam Micol 2009;26:223-7
  • Mistro S, Maciel Ide M, de Menezes RG, et al. Does lipid emulsion reduce amphotericin B nephrotoxicity? A systematic review and meta-analysis. Clin Infect Dis 2012;54:1774-7
  • Gubbins PO, Amsden JR. Drug–drug interactions of antifungal agents and implications for patient care. Expert Opin Pharmacother 2005;6:2231-43
  • Luna B, Drew RH, Perfect JR. Agents for treatment of invasive fungal infections. Otolaryngol Clin North Am 2000;33:277-99
  • Clancy CJ, Nguyen MH. Long-term voriconazole and skin cancer: is there cause for concern? Curr Infect Dis Rep 2011;13:536-43
  • Kethireddy S, Andes D. CNS pharmacokinetics of antifungal agents. Expert Opin Drug Metab Toxicol 2007;3:573-81
  • Black KE, Baden LR. Fungal infections of the CNS: treatment strategies for the immunocompromised patient. CNS Drugs 2007;21:293-318
  • Riddell JT, Comer GM, Kauffman CA. Treatment of endogenous fungal endophthalmitis: focus on new antifungal agents. Clin Infect Dis 2011;52:648-53
  • Fisher JF, Sobel JD, Kauffman CA, et al. Candida urinary tract infections – treatment. Clin Infect Dis 2011;52(Suppl 6):S457-66